AU758464B2 - HIV nuclear localization inhibitors - Google Patents

HIV nuclear localization inhibitors Download PDF

Info

Publication number
AU758464B2
AU758464B2 AU89054/98A AU8905498A AU758464B2 AU 758464 B2 AU758464 B2 AU 758464B2 AU 89054/98 A AU89054/98 A AU 89054/98A AU 8905498 A AU8905498 A AU 8905498A AU 758464 B2 AU758464 B2 AU 758464B2
Authority
AU
Australia
Prior art keywords
compound
branched
straight
hiv
methyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU89054/98A
Other versions
AU8905498A (en
Inventor
Michael Bukrinsky
David Callaway
Omar K Haffar
Senliang Pan
Peter Ulrich
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ferring BV
Original Assignee
Picower Institute for Medical Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Picower Institute for Medical Research filed Critical Picower Institute for Medical Research
Publication of AU8905498A publication Critical patent/AU8905498A/en
Application granted granted Critical
Publication of AU758464B2 publication Critical patent/AU758464B2/en
Assigned to CYTOKINE PHARMASCIENCES, INC. reassignment CYTOKINE PHARMASCIENCES, INC. Alteration of Name(s) in Register under S187 Assignors: PICOWER INSTITUTE FOR MEDICAL RESEARCH, THE
Assigned to FERRING B.V. reassignment FERRING B.V. Alteration of Name(s) in Register under S187 Assignors: CYTOKINE PHARMASCIENCES, INC.
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms

Abstract

There is disclosed a genus of compounds that have anti-HIV infection therapeutic activity and inhibit nuclear localization of the HIV preintegration complex.

Description

WO 99/09014 PCT/US98/16814 HIV NUCLEAR LOCALIZATION INHIBITORS Technical Field of the Invention The present invention provides a genus of compounds and pharmaceutical compositions that have anti-HIV anti-infective therapeutic activity and that inhibit nuclear localization of the HIV preintegration complex.
Background of the Invention In the past decade, infection with the human immunodeficiency virus-type 1 (HIV-1) has reached pandemic proportions. In addition to the overwhelming increase in the number of people infected with HIV-1 in sub-Saharan Africa, there has been a significant increase in new infections in Europe and North America. Of equal concern is the emergence of HIV-1 in Southeast Asian countries such as Thailand and Malaysia. Based on the current rate of infections, it is estimated that Southeast Asia may, in the near future, surpass Africa as the hot spot of the world. Therefore, infection with HIV-1 and development of AIDS proceeds unabated. In the absence of a protective vaccine, post-infection therapy is the only management tool available to health care providers.
The identification of long-term non-progressors strongly suggested that therapy for HIV-1 infection may delay the onset of disease following infection. To date, the principal targets for HIV-1 therapy have been the viral enzymes, reverse transcriptase (RT) and protease, that are important for the virus life cycle. Inhibitors of either of these enzymes successfully reduced the virus load in patients, resulted in increased CD4' T lymphocyte subsets and have become commercially available drugs for HIV infection treatment. Both of these end points have been shown to be good correlates for positive prognosis. Importantly, combination therapies utilizing RT inhibitors together with protease inhibitors in a variety of regimens resulted in reduction of the circulating virus in the blood to below detectable levels. These clinical results showed that maintenance therapy for HIV-1 infection and AIDS is achievable.
However, emergence of virus isolates resistant to the applied anti-viral drugs, as well as cross-resistance to multiple drugs within a class of inhibitors is predicted to limit the application of combination therapy. These results strongly indicated the need for continued novel drug development, and continued identification of novel targets, other than the virus enzymes.
Human immunodeficiency virus type-1 and other lentiviruses infect non-dividing terminally differentiated cells, such as primary macrophages (Gendelman et al., J. Virol. 58:67- 74, 1986: Gartner et al., Science 233:215-219. 1986), primary blood dendritic cells (Langhoff et al., Proc. Natl. Acad. Sci. USA 88:998-8002. 1991), and epidermal Langerhan's cells (Ramazzotti et al., Immunology 85:94-98, 1995). This is facilitated by active importation of the HIV-1 preintegration complex (PIC), which incorporates the viral genome, across the intact nuclear envelope of the non-dividing cell (Bukrinsky et al.. Proc. Natl. Acad. Sci. USA WO 99/09014 PCT/US98/16814 89:6580-6584, 1992: Bukrinsky et al., Nature 365:666-669, 1993; and von Schwedler et al., Proc. Natl. Acad. Sci. USA 91:6992-6996, 1994). In addition, HIV-1 can establish productive infection in activated primary T cells at all steps of the cell cycle, prior to and including the M phase, when dissolution of the nuclear envelope occurs. Thus, active nuclear importation obviates the requirement for cell division, thus allowing HIV-1 to infect non-proliferating as well as proliferating cells (Lewis et al., EMBO J. 11:3053-3058, 1992). the usual targets of retroviruses (Roe et al.. EMBO J. 12:2099-2108, 1993; and Lewis and Emerman. .1 Virol.
68:510-516. 1994).
In addition to the viral genomic RNA, the PIC is composed of the gag-derived matrix antigenprotein nucleocapsid protein reverse transcriptase integrase (IN), and viral protein (vpr). Reverse transcription and production of the nascent cDNA is completed in context of the PIC in the cytoplasm of the infected target cell, prior to nuclear entry. It was recently shown (Gallay et al.. J. Virol. 70:1027-1032, 1996; and Popov et al., Proc. Natl. Acad. Sci. USA 93:11859-11864. 1996) that the PIC of HIV-1 associates with karyopherins, the cellular proteins involved in active nuclear importation (reviewed in Adam, Trends Cell Biol. 5:189-191, 1995). Karyopherin a binds to target proteins via their nuclear localization sequence (NLS), while karyopherin P mediates docking of the karyopherin atarget protein complex to nuclear pore structures (Radu et al., Proc. Natl. Acad Sci. USA 92:1769-1773, 1995; Moroianu et al., Proc. Natl. Acad. Sci USA 92:2008-2011, 1995; Gorlich et al., Nature (London) 377:246-248, 1995; Adam and Gerace, Cell 66:837-847, 1991; Gbrlich and Mattaj, Science 271:1513-1518, 1996; and Hurt, Cell 84:509-515. 1996).
HIV-1 matrix antigen protein contains one defined (K 2 6 KKYK) and one putative
(K'
i °SKKK) NLS, and represents a major karyophilic structure within the PIC (Bukrinsky et al., Nature 365:666-669. 1993; von Schwedler et al., Proc. Natl. Acad Sci. USA 91:6992-6996, 1994; Gallay et al., J. Virol. 70:1027-1032, 1996; and Bukrinsky et al. Proc. Natl. Acad. Sci.
USA 90:6125-6129, 1993). Synthetic peptides encompassing either of the two MA NLS bound both identified human karyopherin a present in B cell and T cell lysates (Nadler et al.. J. Biol.
Chem. 272, 4310-4315, 1997). Mutations in the KKKYK NLS of MA, alone or in combination with the deletion of Vpr, reduced nuclear importation of the HIV-1 PIC and inhibited infection of primary macrophage cultures (von Schwedler et al., Proc. Natl. Acad.
Sci. USA 91:6992-6996. 1994; Heizinger et al., Proc. Natl. Acad. Sci. USA 91:7311-7315, 1992), as well as growth-arrested T cells (Bukrinsky et al.. Nature 365:666-669, 1993) and CD4 -HeLa cell cultures (Emerman et al., Nature (London) 369:107-108, 1994). Single amino acid substitutions within the KKKYK NLS also reduced binding of the HIV-1 PIC to yeast karyopherin a in vitro (Popov et al., Proc. Natl. Acad Sci. USA 93:11859-11864. 1996), thus providing a link between binding of PIC to karyopherin a, nuclear import, and viral replication in non-dividing cells.
WO 99/09014 PCT/US98/16814 Summary of the Invention The present invention provides a compound having the formula 1: 0 0
Z
A
A
SR
wherein A is independently a straight or branched C .6 alkyl, a straight or branched C 26 alkenyl or a C 16 alkoxy; Y is -S-A wherein A is independently defined above; and Z and X are independently H, -(CH 2 )n-NH wherein n is an integer from 0 to 6. a straight or branched Ci-6 alkyl, a straight or branched C2- 6 alkenyl or a Ci.
6 alkoxy. Preferably, A is methyl, X is -NH 2 and Z is H or amino. The present invention further provides a pharmaceutical composition comprising a compound from formula 1 in a pharmaceutically acceptable carrier.
The invention further provides a process for synthesizing a compound of formula I, comprising the steps of: providing a solution of acetyl chloride in a short chain alcohol; adding to the solution a substituted 6-halogen-methylmercaptopyrimidine and a 3,5-dialkylaniline to form a mixture; refluxing the mixture to join the aniline derivative to the 6 position of the pyrimidine derivative; and drying the mixture to obtain a solid final product according to formula I.
Preferably, the substituted 6 -halogen-methylmercaptopyrimidine is 4-amino-6-chloro-2methylmercaptopyrimidine and the 3.5-dialkylaniline is Brief Description of the Drawings Figure 1 shows a graph comparing, in an assay of anti-HIV activity in macrophage cultures, anti-HIV therapeutic activity of inventive compound 53 (2-methylmercapto-4-amino- 6-(3',5'-diacetylphenyl) amino-pyrimidine) with a structurally similar compound ("cnih0294") that differs from compound 53 of the present invention by having a positive charge in the pyrimidine moiety and lacking a stipulated sulfur group substituted to the pyrimidine moiety. The assay measures reverse transcriptase activity in the infected macrophage culture supernatants as a measure of virus production. These data can be directly correlated to efficacy treating HIV infection. These data show that inventive compound 53 was more efficacious that structurally similar compound 2.
Figure 2 shows a graph comparing anti-HIV therapeutic activity, in an assay of anti- HIV activity in macrophage cultures, of inventive compound 62 (2-methylmercapto-6-(3',5'diacetyiphenyl) amino-pyrimidine) with a structurally similar compound ("cni-h0294") that WO 99/09014 PCT/US98/16814 differs from compound 62 of the present invention by having a positive charge in the pyrimidine moiety and lacking a stipulated sulfur group substituted to the pyrimidine moiety.
The assay measures reverse transcriptase activity in the infected macrophage culture supernatants as a measure of virus production. These data can be directly correlated to efficacy treating HIV infection. These data show that inventive compound 62 was an efficacious anti- HIV anti-infective agent.
Figure 3 shows a further analysis of therapeutic efficacy of compound 62 in activated (anti-CD3 and anti-CD28 monoclonal antibodies) peripheral blood mononuclear cell (PBMC) cultures infected with HIV-1 virus and treated with different concentrations of compound 62 (jpM). The assay measures p24 as an index of viral replication and can be directly correlated to efficacy in treating HIV infection. These data show anti-viral efficacy of compound 62 in a dose-response fashion.
Figure 4 shows that compound 62 also inhibited virus replication in PBMC from a HIV-1 infected individual when the PBMCs were activated in vitro with anti-CD3 mAb.
PBMCs from a seropositive individual were collected and depleted of CD8' T lymphocytes as described above. Cells were suspended in culture medium and activated with anti-CD3 mAb (1 g/ml). After 6-10 days virus production was evaluated by measuring levels of p24 in the culture supernatants and comparing treated to untreated cultures. Figure 4 shows a doseresponse relationship for compound 62 ("cni-h6297") under the foregoing experimental conditions in this predictive assay of HIV anti-infective properties.
Detailed Description of the Invention The present invention provides a compound having the formula I: O 0
Z
A A
R-
wherein A is independently a straight or branched Ci-6 alkyl, a straight or branched C 2 6 alkenyl or a Ci 1 6 alkoxy; Y is -S-A wherein A is independently defined above: and Z and X are independently H, -(CH 2 )n-NH 2 wherein n is an integer from 0 to 6, a straight or branched Ci-6 alkyl, a straight or branched C2- 6 alkenyl or a C 1 -6 alkoxy. Preferably, A is methyl, X is -NH 2 and Z is H or amino. The present invention further provides a pharmaceutical composition comprising a compound from formula I in a pharmaceutically acceptable carrier.
The invention further provides a process for synthesizing a compound of formula I, comprising the steps of: providing a solution of acetyl chloride in a short chain alcohol; WO 99/09014 PCT/US98/16814 adding to the solution a substituted 6 -halogen-methylmercaptopyrimidine and a to form a mixture; refluxing the mixture to join the aniline derivative to the 6 position of the pyrimidine derivative; and drying the mixture to obtain a solid final product according to formula I.
Preferably, the substituted 6-halogen-methylmercaptopyrimidine is 4-amino-6-chloro-2methylmercaptopyrimidine and the 3,5-dialkylaniline is The present invention provides an improvement in the design of small organic molecules that are effective for inhibiting HIV infection by creating an integral sulfurcontaining substituent (see in formula The presence of this substituent, not disclosed or suggested in alternative HIV inhibitors, has provided compound characteristics of improved cellular absorption and, as a result, improved potency not disclosed or suggested by the structures of the alternative HIV preintegration complex inhibitors.
The inventive compounds were active to inhibit receptor-mediated nuclear importation in the infection of peripheral blood mononuclear cell (PBMC) cultures. The compound, cnih0294, is thought to interact with the HIV-1 PIC (preintegration complex) by forming partial Schiff bases with adjacent lysine residues in the MA NLS (Popov et al., Proc. Natl. Acad. Sci.
USA 93:11859-11864, 1996; Dubrovsky et al., Molec. Med. 1:217-230. 1995), contains a positive charge in its substituted pyrimidine moiety that may serve to limit its cellular bioavailability and it oral absorption characteristics. It has been previously shown that cnih0294 interferes with the association of the HIV PIC with the yeast karyopherin ao (Popov et al., Proc. Natl. Acad. Sci. USA 93:11859-11864, 1996), and effectively inhibits infection of primary macrophage cultures with the macrophage tropic isolate HIVADA (IC 5 o=10nM-50nM) (Dubrovsky et al., Molec. Med. 1:217-230, 1995).
It should be noted that in order for a therapeutic agent to be effective as a PIC inhibitor.
it must act intracellularly. Thus, potency is directly related to cellular bioavailability.
Moreover, the preferred route of administration to treat HIV infection on a chronic basis is oral to increase patient compliance. Therefore, it is highly desirable to have an HIV anti-infective compound be administered orally and have high oral bioavailability.
Compound Synthesis The exemplar, compound. 2-methylmercapto-4-amino-6-(3',5'-diacetylphenyl) aminopyrimidine (compound 53) was synthesized. There are two methods for synthesizing compound 53. The first method starts by adding acetyl chloride (0.8 ml, 11 mmol) to 60 ml absolute ethanol. The mixture was stirred for 15 minutes to form a hydrochloric ethanol solution. Then, 1.75 g (10 mmol) of 4-amino-6-chloro-2-methylmercaptopyrimidine (Aldrich) and 3,5-diacetylaniline (1.8 g, 10 mmol) (Aldrich) were added in sequence. The reaction mixture was refluxed for 24 hours and eventually turned a brown color. The ethanol was evaporated to dryness and 20 ml CHCl 3 was added to the residue (a gray solid residue) and separated. The gray solid was filtered to give 1.7 g crude product after drying. A TLC (thin WO 99/09014 PCT/US98/16814 layer chromatography analysis showed that the unreacted starting material stayed in the CHC13 layer. Approximately 0.5 g of crude product was recrystalized in methanol to give 370 mg of analytical pure sample (yield 39.8%).
There is a second method to product compound 53. Specifically, HCI (90 Pl) was added to a mixture containing 4-amino-6-chloro-2-methylmercapto pyridine (1 mmol) and diacetvlanaline in 5 ml H 2 0 (1.1 mmol). The reaction mixture was heated at 90-100 °C for 1 hour, and then cooled down in an ice bath. Two ml of 1 N KOH was added to neutralize the acid. The mixture was stirred for 10 minutes, a precipitate formed and then the precipitate was filtered out to give 285 mg dry, pale brown crude product (yield A TLC analysis (MeOH:CH 2 Cl 2 10:0.6) showed only one spot for a pure product. Recrystalization from a methoxyethanol solution provided 267 mg of analytically pure product (yield The recrystalized product was dried under vacuum (78 with a melting point of 264.8 0 C-268.6 oC.
The analytical specifications of compound 53 are molecular weight 316.4 and Ci 5
H
1 6
N
4 0 2 S. Additionally, 'HNMR (DMSO-d6, 270 MHz): 6 2.46 3H, SMe), 2.6 6H, 2
COCH
3 5.52 1H, 6.52 (br s, 2H, NH 2 8.01 1H, Ar-H). 8.45 2H. Ar-H), 9.41 1 H, NH). Further analysis of the final product compared the found elemental analysis versus the calculated elemental analysis: Calculated: C56.94 H5.10 N17.17 S10.14 Found: C56.84 H4.99 N17.55 S10.18 Illustrative compound 62 (2-methylmercapto-6-(3',5'-diacetylphenyl) aminopyrimidine) was synthesized by mixing 803 mg (5 mmol) 2-methylmercapto-4chloropyrimidine and 886 mg (5 mmol) 3,5-diacetylaniline in 20 ml of water. In addition, 0.42 ml concentrated HCI was added. This reaction mixture was heated at 90-100 °C for 4 hours, and then cooled down in an ice bath. The cooled mixture had 5 ml of 1N KOH added to neutralize the acid pH from the HCI. The mixture was stirred for 10 minutes in an ice bath to form a precipitate. The precipitate was filtered out to give 1.44 g dry pale brown crude product. A thin layer chromatography (TLC) analysis in methanol:CH 2 Cl 2 (1:25) showed only one spot. The crude precipitate was recrystalized from methoxyethanol and dried under vacuum to give 1.42 grams of pure 2-methylmercapto-4-(3',5'-diacetylphenyl) aminopyrimidine (compound 62) with an overall yield of 94%. The NMR analysis found the formula C IH 15
N
3 02S with a molecular weight of 301.38.
calculated: C 59.78 H 5.02 N 13.94 S 10.64 found: C 59.32 H 4.80 N 13.81 S 10.34 Pharmaceutical Formulation The inventive pharmaceutical complex or inventive pharmaceutical combination can be administered to a patient either by itself (complex or combination) or in pharmaceutical compositions where it is mixed with suitable carriers and excipients. The inventive compound or pharmaceutical composition can be administered parenterally, such as by intravenous WO 99/09014 PCT/US98/16814 injection or infusion, intraperitoneal injection, subcutaneous injection, or intramuscular injection. The inventive compound or pharmaceutical composition can be administered orally or rectally through appropriate formulation with carriers and excipients to form tablets, pills, capsules. liquids, gels, syrups, slurries, suspensions and the like. The inventive compound or pharmaceutical composition can be administered topically, such as by skin patch, to achieve consistent systemic levels of active agent. The inventive compound or pharmaceutical composition is formulated into topical creams, skin or mucosal patches, liquids or gels suitable to topical application to skin or mucosal membrane surfaces. The inventive compound or pharmaceutical composition can be administered by inhaler to the respiratory tract for local or systemic treatment of HIV infection.
The dosage of the inventive compound or pharmaceutical composition suitable for use with the present invention can be determined by those skilled in the art from this disclosure.
The pharmaceutical composition will contain an effective dosage (depending upon the route of administration and pharmacokinetics of the active agent) of the inventive compound or pharmaceutical composition and suitable pharmaceutical carriers and excipients, which are suitable for the particular route of administration of the formulation oral, parenteral, topical or by inhalation). The active compound is mixed into the pharmaceutical formulation by means of mixing, dissolving, granulating, dragee-making, emulsifying, encapsulating, entrapping or lyophilizing processes. The pharmaceutical formulations for parenteral administration include aqueous solutions of the active complex or combination in watersoluble form. Additionally, suspensions of the active compound may be prepared as oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose. sorbitol. or dextran. The suspension may optionally contain stabilizers or agents to increase the solubility of the complex or combination to allow for more concentrated solutions.
Pharmaceutical formulations for oral administration can be obtained by combining the active compound with solid excipients, such as sugars lactose, sucrose, mannitol or sorbitol), cellulose preparations starch, methyl cellulose, hydroxypropylmethyl cellulose, and sodium carboxymethyl cellulose), gelatin, gums, or polyvinylpyrrolidone. In addition, a desintegrating agent may be added, and a stabilizer may be added.
Example 1 This example illustrates several in vitro experiments in predictive models of treatment of HIV infection to show the therapeutic utility of the inventive compounds. Macrophages, isolated and purified as described by Schumandtmanesalla et al. (Virology, 1997)) were infected with HIV-I at a multiplicity adjusted according to p24 content (10 ng p24 per 106 cells). Compound (53 in Figure 1 or 62 in Figure 2) was added at different concentrations. In WO 99/09014 PCT/US98/16814 addition, positive control compound 2 (called "CNI-H294" in Figure 1 and "cni-h0294" in Figure 2) was added at the concentrations indicated. After a two hour incubation for viral adsorption, excess viruses were washed away, and the cells were incubated for additional indicated periods prior to analysis. RT, or reverse transcriptase activity, was measured by standard techniques in 7-11 days.
Figure 1 shows a graph comparing anti-HIV therapeutic activity of inventive compound 53 with a structurally similar compound ("compound having a positive charge in the pyrimidine moiety and lacking a required sulfur group substituted to the pyrimidine moiety in an assay of anti-HIV activity in H9 cell cultures. The assay measures reverse transcriptase activity in the infected macrophage culture supematants as a measure of virus production.
These data can be directly correlated to efficacy treating HIV infection. These data show that inventive compound 53 was more efficacious that structurally similar compound 2.
Figure 2 shows a graph comparing anti-HIV therapeutic activity of inventive compound 62 with a structurally similar compound ("cni-h0294") having a positive charge in the pyrimidine moiety and lacking a required sulfur group substituted to the pyrimidine moiety in an assay of anti-HIV activity in macrophage cultures. The assay measures reverse transcriptase activity in the infected H9 cell culture supernatants as a measure of virus production. These data can be directly correlated to efficacy treating HIV infection. These data show that inventive compound 62 was an efficacious anti-HIV anti-infective agent.
Example 2 This example illustrates that compound 62 inhibited HIV-1 virus replication in acutely infected PBMC cultures activated with anti-CD3 and anti-CD28 monoclonal antibodies (Figure Peripheral blood mononuclear cells were isolated from an uninfected individual and depleted of CD8' T lymphocytes using a CD8-specific monoclonal antibody, according to the procedure described by Smithgall et al., J. Immunol. 156:2324-2330, 1996. Briefly, the procedure substitutes separation with magnetic beads for complement-mediated lysis of antibody-bound cells. The remaining PBMC fractions were suspended in RPMI culture medium supplemented with 10% heat-inactivated human serum at 2x106 cells/200pl. Cells were activated with anti-CD3 mAb (lplg/ml) together with anti-CD28 mAb (1 pg/ml) in the presence of various concentrations of compound 62. This form of cell activation specifically targets CD3' T lymphocytes in the population.
Cells were pretreated with antibody and test compound for 2-3 hours prior to addition of the virus inoculum. The virus used in this experiment, HIV-1MI, is a patient-derived isolate, and was used at an approximate multiplicity of infection (MOI) =5 TCID 50 After 2 hr incubation for adsorption of virus, the cells were washed free of the inoculum, and then resuspended in 200 ml of culture medium supplemented with anti-CD3 and anti-CD28 mAbs together with varyious concentrations of compound 62 (to show a dose-response relationship).
Cells were then placed into a U-bottom 96 well culture plate in 4-6 replicates at 1.5x10 Q:\OPEU\MS\226236O.I1.dO- 1 7YO1I03 -9cells/well. Virus production was measured on day 6-10 following infection using p24 production as an end point. The p24 antigen capture assay was performed according to the manufacturer's recommendations.
The data presented in Figure 3 show a dose-response relationship at 0, 0.01 j.±M and 5: LiM concentrations of compound 62 ("CNI-H62 97") when using p24 as a measure of virus concentration.
S, ***eCompound 62 also inhibited virus replication in PBMC from an HIV- I infected individual when the PBMCs were activated in vitro with anti-CD3 mAb. PBMCs from a seropositive individual were collected and depleted of CD 8' T lymphocytes as described 10 above. Cells were suspended in culture medium and activated with anti-CD3 mAb (Ilpg/ml).
After 6-10 days virus production was evaluated by measuring levels of p24 in the culture supernatants and comparing treated to untreated cultures. Figure 4 shows a dose-response relationship for compound 62 ("CNI-H6297") under the foregoing experimental conditions in this predictive assay of HIV anti-infective properties.
0000 Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge in Australia.

Claims (11)

1. A compound having the formula I: O 0 Z 0I 5 wherein A is independently a straight or branched C.-6 alkyl, a straight or branched C 2 -6 alkenyl or a C 1 -6 alkoxy; Y is -S-A wherein A is independently defined above; and Z and X are independently H, -(CH 2 )n-NH 2 wherein n is an integer from 0 to 6, a straight or branched C 1 6 alkyl, a straight or branched C2- 6 alkenyl or a C 1 6 alkoxy.
2. The compound of claim 1 wherein A is methyl, X is -NH2 and Z is H.
3. The compound of claim 1 wherein A is methyl, X is -NH 2 and Z is amino.
4. A pharmaceutical composition comprising a compound of claim 1 in a S pharmaceutically acceptable carrier.
5. The pharmaceutical composition of claim 4 wherein A is methyl, X is -NH 2 and SZ is H. 15
6. The pharmaceutical composition of claim 4 wherein A is methyl, X is -NH 2 and Z is amino.
7. A process for synthesizing a compound of formula I, comprising the steps of: S(a) providing a solution of acetyl chloride in a short chain alcohol; adding to the solution a substituted halogen-methylmercaptopyrimidine and a 20 dialkylaniline to form a mixture; refluxing the mixture to join the aniline derivative to the pyrimidine derivative; and drying the mixture to obtain a solid final product according to formula I.
8. The process of claim 7 wherein the substituted 6-halogen- methylmercaptopyrimidine is 4-amino-6-chloro-2-methylmercaptopyrimidine and the dialkylaniline is
9. A method for treating HIV infection, comprising administering an effective amount of a compound having the formula I: O O Z A A 0 wherein A is independently a straight or branched Ci. 6 alkyl, a straight or branched C 2 6 alkenyl QAOPER/JMS\2262360.017,d,.-j 7/01/03 -11 I- or a C 1 6 alkoxy; Y is -S-A wherein A is independently defined above; and Z and X are independently H, -(CH 2 2 wherein n is an integer from 0 to 6, a straight or branched C1- alkyl, a straight or branched C 2 6 alkenyl or a C 1 6 alkoxy.
The method of claim 9 wherein A is methyl, X is -NH 2 and Z is H-.
11. The method of claim 9 wherein A is methyl, X is -NH, and Z is amino. Dae hs.7t ayo Th ioe nttt frMdclRsac Byis aen ttmy Daie *.li Cv 0
AU89054/98A 1997-08-15 1998-08-13 HIV nuclear localization inhibitors Ceased AU758464B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/912,076 US5808068A (en) 1997-08-15 1997-08-15 HIV nuclear localization inhibitors
US08/912076 1997-08-15
PCT/US1998/016814 WO1999009014A1 (en) 1997-08-15 1998-08-13 Hiv nuclear localization inhibitors

Publications (2)

Publication Number Publication Date
AU8905498A AU8905498A (en) 1999-03-08
AU758464B2 true AU758464B2 (en) 2003-03-20

Family

ID=25431342

Family Applications (1)

Application Number Title Priority Date Filing Date
AU89054/98A Ceased AU758464B2 (en) 1997-08-15 1998-08-13 HIV nuclear localization inhibitors

Country Status (8)

Country Link
US (1) US5808068A (en)
EP (1) EP1012146B1 (en)
JP (1) JP4475487B2 (en)
AT (1) ATE245983T1 (en)
AU (1) AU758464B2 (en)
CA (1) CA2300424C (en)
DE (1) DE69816835T2 (en)
WO (1) WO1999009014A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6297253B1 (en) * 1996-10-15 2001-10-02 The Picower Institute For Medical Research Compounds and methods of use to treat infectious diseases
CA2475189C (en) * 2003-07-17 2009-10-06 At&T Corp. Method and apparatus for window matching in delta compressors
US20050203150A1 (en) * 2004-03-09 2005-09-15 Haffar Omar K. Compounds, compositions and methods for inhibiting or treating HIV-1

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0673022A (en) * 1992-08-27 1994-03-15 Kumiai Chem Ind Co Ltd Pyrimidine or triazine derivative and herbicide

Also Published As

Publication number Publication date
AU8905498A (en) 1999-03-08
EP1012146A1 (en) 2000-06-28
DE69816835T2 (en) 2004-04-22
WO1999009014A1 (en) 1999-02-25
JP4475487B2 (en) 2010-06-09
EP1012146A4 (en) 2002-12-04
CA2300424A1 (en) 1999-02-25
ATE245983T1 (en) 2003-08-15
JP2001515069A (en) 2001-09-18
US5808068A (en) 1998-09-15
EP1012146B1 (en) 2003-07-30
CA2300424C (en) 2009-04-07
DE69816835D1 (en) 2003-09-04

Similar Documents

Publication Publication Date Title
Selvam et al. Synthesis and anti-HIV activity of 4-[(1, 2-dihydro-2-oxo-3H-indol-3-ylidene) amino]-N (4, 6-dimethyl-2-pyrimidinyl)-benzene sulfonamide and its derivatives
Tanaka et al. Synthesis and anti-HIV activity of 2-, 3-, and 4-substituted analogs of 1-[(2-hydroxyethoxy) methyl]-6-(phenylthio) thymine (HEPT)
US5703086A (en) Compounds and methods of use to derivatize neighboring lysine residues in proteins under physiological conditions
US20060094755A1 (en) Novel quinoline-based metal chelators as antiviral agents
JPH04230280A (en) Aminobenzodiazepine and drug containing same
WO1997038999A9 (en) Acridone-derived compounds useful as antineoplastic and antiretroviral agents
AU759210B2 (en) HIV matrix protein tyrosine position 29 pocket binders
AU758464B2 (en) HIV nuclear localization inhibitors
US20060148849A1 (en) Quinoline derivatives, process of synthesis and medicaments containing these derivatives
Al-Abed et al. Inhibition of HIV-1 nuclear import via Schiff base formation with arylene bis (methylketone) compounds
EP0843663B9 (en) 4-aryl-thio-pyridin-2(1h)-ones, drugs containing same, and uses thereof for treating hiv-related diseases
JP2021500401A (en) Wide spectrum antiviral compositions and methods
US5574040A (en) Pyrimidine compounds and methods of use to derivatize neighboring lysine residues in proteins under physiologic conditions
Michne et al. Keto/enol epoxy steroids: A new structural class of hiv-1 tat inhibitors
US20170267726A1 (en) Non-immunosuppressive cyclosporin derivatives as antiviral agents
CN111995622B (en) Compound and use thereof
JP2007506788A (en) HCV infection inhibitors and uses thereof
EP2163553A1 (en) Arene connected polyamine macroring derivatives, preparation methods and pharmaceutical uses thereof
Kasralikar et al. Month 2018 Design and Synthesis of Novel 1, 2, 3-triazolyl-pyrimidinone Hybrids as Potential Anti-HIV-1 NNRT Inhibitors
CN115403625A (en) S-DACOs non-nucleoside reverse transcriptase inhibitor derivative and application thereof
WO2005016343A1 (en) Novel anti-viral agents based upon derivatives of the aromatic heterocycle phenanthridine
US6414004B1 (en) 3-substituted 5-aryl-4-isoxazolecarbonitriles having antiviral activity
CA2525550C (en) Acridone-derived compounds useful as antiretroviral agents
CA2250863C (en) Acridone-derived compounds useful as antineoplastic and antiretroviral agents
US20050203150A1 (en) Compounds, compositions and methods for inhibiting or treating HIV-1

Legal Events

Date Code Title Description
DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS: ADD INVENTORS DAVID CALLAWAY AND PETER ULRICH

FGA Letters patent sealed or granted (standard patent)